Full Text

advertisement
Hair-Cell Mechanotransduction Persists in TRP Channel Knockout
Mice
The Harvard community has made this article openly available.
Please share how this access benefits you. Your story matters.
Citation
Wu, Xudong, Artur A. Indzhykulian, Paul D. Niksch, Roxanna M.
Webber, Miguel Garcia-Gonzalez, Terry Watnick, Jing Zhou,
Melissa A. Vollrath, and David P. Corey. 2016. “Hair-Cell
Mechanotransduction Persists in TRP Channel Knockout Mice.”
PLoS ONE 11 (5): e0155577. doi:10.1371/journal.pone.0155577.
http://dx.doi.org/10.1371/journal.pone.0155577.
Published Version
doi:10.1371/journal.pone.0155577
Accessed
October 2, 2016 11:25:06 AM EDT
Citable Link
http://nrs.harvard.edu/urn-3:HUL.InstRepos:27662191
Terms of Use
This article was downloaded from Harvard University's DASH
repository, and is made available under the terms and conditions
applicable to Other Posted Material, as set forth at
http://nrs.harvard.edu/urn-3:HUL.InstRepos:dash.current.terms-ofuse#LAA
(Article begins on next page)
RESEARCH ARTICLE
Hair-Cell Mechanotransduction Persists in
TRP Channel Knockout Mice
Xudong Wu1☯, Artur A. Indzhykulian1☯, Paul D. Niksch1, Roxanna M. Webber1,
Miguel Garcia-Gonzalez2, Terry Watnick2, Jing Zhou3, Melissa A. Vollrath1,4, David
P. Corey1*
a11111
1 Department of Neurobiology, Harvard Medical School and Howard Hughes Medical Institute, Boston,
Massachusetts, United States of America, 2 Department of Medicine, Division of Nephrology, University of
Maryland, Baltimore, Maryland, United States of America, 3 Renal Division, Department of Medicine,
Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of
America, 4 Department of Physiology, McGill University Montréal, Québec, Canada
☯ These authors contributed equally to this work.
* dcorey@hms.harvard.edu
Abstract
OPEN ACCESS
Citation: Wu X, Indzhykulian AA, Niksch PD,
Webber RM, Garcia-Gonzalez M, Watnick T, et al.
(2016) Hair-Cell Mechanotransduction Persists in
TRP Channel Knockout Mice. PLoS ONE 11(5):
e0155577. doi:10.1371/journal.pone.0155577
Editor: Bernd Sokolowski, University of South
Florida, UNITED STATES
Received: March 4, 2016
Accepted: May 1, 2016
Published: May 19, 2016
Copyright: © 2016 Wu et al. This is an open access
article distributed under the terms of the Creative
Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any
medium, provided the original author and source are
credited.
Data Availability Statement: All relevant data are
within the paper and its Supporting Information files.
Funding: This work used reagents provided by the
Baltimore Polycystic Kidney Disease Research and
Clinical Core Center (P30DK090868). Supported by
NIH grant DC000304 to DPC, and R01DK51050,
R01DK53357 and R01DK09953201 to JZ. The
funders had no role in study design, data collection
and analysis, decision to publish, or preparation of
the manuscript.
Competing Interests: The authors have declared
that no competing interests exist.
Members of the TRP superfamily of ion channels mediate mechanosensation in some
organisms, and have been suggested as candidates for the mechanotransduction channel
in vertebrate hair cells. Some TRP channels can be ruled out based on lack of an inner ear
phenotype in knockout animals or pore properties not similar to the hair-cell channel. Such
studies have excluded Trpv4, Trpa1, Trpml3, Trpm1, Trpm3, Trpc1, Trpc3, Trpc5, and
Trpc6. However, others remain reasonable candidates. We used data from an RNA-seq
analysis of gene expression in hair cells as well as data on TRP channel conductance to
narrow the candidate group. We then characterized mice lacking functional Trpm2, Pkd2,
Pkd2l1, Pkd2l2 and Pkd1l3, using scanning electron microscopy, auditory brainstem
response, permeant dye accumulation, and single-cell electrophysiology. In all of these
TRP-deficient mice, and in double and triple knockouts, mechanotransduction persisted.
Together with published studies, these results argue against the participation of any of the
33 mouse TRP channels in hair cell transduction.
Introduction
Sound conducted to the cochlea causes the movement of stereocilia on hair cells, the receptor
cells of the inner ear. Sub-micron deflection of the bundle of stereocilia on a hair cell opens ion
channels in microseconds, allowing influx of cations and the generation of a receptor potential
[1,2]. Although a great deal is known about the ultrastructure and molecular mechanics of the
mechanotransdution apparatus, the molecular identity of the transduction channel has been
uncertain.
The physiological properties of this elusive channel provide a fingerprint for screening candidates. The transduction channel is a nonselective cation channel with high permeability to
Ca2+ (PCa/PNa = 5–20) [3,4,5]. Although many divalent cations are permeant, they are also
channel blockers: the channel can be blocked by Ca2+ (IC50 = 1 mM), Mg2+, La3+ (4 μm), and
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
1 / 22
TRP Channels in Hair Cell Mechanotransduction
Gd3+ (3 μm) [6,7,8]. The single channel conductance varies considerably, ranging from about
80 to 150 pS in 2–3 mM extracellular Ca2+, and is roughly twice that in low Ca2+ [6,9,10,11,12].
Some organic cations are also permeant blockers, such as amiloride (IC50 = 50 μm [13]), the
fluorescent dye FM1-43 (2 μm [14]) and the antibiotic dihydrostreptomycin (10‐70 μM
[4,15,16]). The block is voltage dependent indicating that these cations block within the pore,
part way along the transmembrane electric field [14,17]. Finally, transduction channels are partially permeable to large organic cations, such as choline and TEA, up to about 12 Å diameter
[8]. The current view of the transduction channel shows a funnel-shaped channel with an
outer vestibule and 12 Å selectivity filter [8,16,18,19]. These properties suggested that members
of the transient receptor potential (TRP) family of ion channels, especially the PKD2 group,
would be good candidates for the transduction channel [20,21].
The timing of gene expression provides additional clues. In mice, vestibular hair cells
become mechanosensitive beginning on embryonic day 17 (E17) Cochlear hair cells show
mechanosensitivity beginning between postnatal day 0 (P0) and P2, in the base and apex
respectively [22,23]. We expect mRNA for the transduction channel gene to appear at or
slightly before these times. Cuajungco et al. [20] analyzed expression of all 33 TRP channels in
a mammalian organ of Corti library, and found 19 TRPs expressed at a single age. Asai et al.
[24] went on to analyze expression of mRNAs for all TRP channels in cochlea using RT–PCR
from whole inner ear tissue, at E17, E18, P0, P2, P4, P6 and P8 [24]. However, they were unable
to distinguish expression in hair cells from that in supporting cells and other surrounding cells,
somewhat limiting the usefulness of the analysis.
Here, we explore TRP channels as candidates for the hair cell transduction channel. We
take advantage of new data on specific gene expression in hair cells at different developmental
time points to narrow the candidates, and further narrow candidates by single-channel conductance and phenotypes in published TRP knockouts. With scanning electron microscopy,
FM1-43 loading and single cell physiology, we investigate transduction in mouse knockouts of
Trpm2, Pkd2, Pkd2l1, Pkd2l2 and Pkd1l3, and in double and triple knockouts of these genes.
We find no substantial deficit in mechanotransduction, ruling out these TRPs. Although many
TRPS are expressed in hair cells and surely have important functions, we argue that none of
the TRP channels are likely candidates for the transduction channel itself.
Results
To understand specific expression of TRP channels in hair cells, we queried an extensive database of gene expression in hair cells during development [25]. In that study, hair cell- and surrounding cell-specific RNA-seq libraries were prepared from FACS-sorted cochlear and
vestibular tissues at E16, P0, P4 and P7. The expression of the TRP channel family for different
cell types and ages is shown in Fig 1. The single-channel conductance, drawn from the literature [26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,
55,56,57,58,59,60,61,62,63,64,65,66,67,68] is also indicated. For reference, S1 Fig shows the
TRP family in mouse arranged phylogenetically.
In narrowing candidates, we felt that an attractive transduction channel candidate should
show at least two-fold enrichment in hair cells compared to surrounding cells, and it should be
expressed in hair cells by the time of transduction onset. It should preferably have a large single
channel conductance (>80 pS) although it is possible that a heteromeric TRP channel might have
higher conductance than either subunit expressed alone. It would be more attractive if expressed
in both auditory and vestibular hair cells [25] and in both inner and outer hair cells in the cochlea
[69]. Knockout mice have been generated for a number of TRP channels, and we excluded all
TRPs that have been reported to have normal hair cell transduction or showed no phenotype that
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
2 / 22
TRP Channels in Hair Cell Mechanotransduction
Fig 1. TRP channel expression in cochlear and vestibular hair cells. (a) Schematic of the sorted cells, redrawn from
Scheffer et al., 2015 [25]. Hair cells (green) and surrounding cells (purple) were collected from the cochlea (dark colors) and
utricle (light colors) at ages E16, P0, P4, and P7. (b) Normalized RNA-seq read counts in hair cells and surrounding cells (colors
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
3 / 22
TRP Channels in Hair Cell Mechanotransduction
as in a). Data are shown for 31 of the 33 members of the TRP channel family in mouse. Mcoln2 and Trpc4 showed negligible
counts in all samples. Data from www.shield.hms.harvard.edu [25].
doi:10.1371/journal.pone.0155577.g001
might be expected from auditory or vestibular deficits. In mice, loss of vestibular function often
leads to circling, spinning or head-bobbing behavior. Such behavior is often the first indication of
an inner-ear phenotype in random or targeted mutagenesis and is usually noted in phenotypic
descriptions. Auditory function is usually tested only if there is a reason to suspect dysfunction,
for instance if vestibular problems are noted. Trpa1, Trpc1, Trpc3, Trpc5, Trpc6, Trpv4, Trpml3
and Trpm1 can be excluded on this basis [70,71,72,73,74]. Similarly, channels with a reported
conductance <80 pS were excluded unless other strong indications were present.
Trpm2 is not required for hair cell transduction
Based on these criteria, we first focused on Trpm2. Its mRNA shows 90-fold enrichment in hair
cells compared to surrounding cells, and it is first expressed at E17 in vestibular system and P0
in cochlea, matching the onset of mechanosensitivity. Expression further increases during
development in both tissues. Although the published conductance of 50–60 pS is somewhat
below our criterion, the expression pattern warranted further investigation.
We therefore generated a conditional Trpm2 knock-out mouse in which exon 21—encoding
the fifth transmembrane and pore domains of Trpm2—is flanked by LoxP sites (Trpm2fl/fl; see
Methods and S2 Fig). Trpm2fl/fl mice were viable and could be bred as homozygotes. For haircell-specific Cre-mediated recombination, we crossed Trpm2fl/fl mice with mice expressing
Cre-recombinase under control of the Gfi1 promoter [75]. PCR from genomic DNA purified
from inner ears of Trpm2fl/fl:Gfi1-Cre+/- mice confirmed the deletion.
Trpm2fl/fl:Gfi1-Cre+ homozygous knockout mice looked similar to their Trpm2fl/+:
Gfi1-Cre+/- heterozygous control littermates, suggesting no gross developmental defects.
Gfi1 is expressed in vestibular hair cells, so Cre recombination should delete Trpm2 in these
cells, however Trpm2-deficient mice showed no obvious vestibular deficit.
To test mechanotransduction in Trpm2-deficient cochlear hair cells, we applied FM1-43
dye [76] to cultured organ of Corti explants. Both control and Trpm2-deleted hair cells accumulated FM1-43, showing similar levels of fluorescence intensity, suggesting that mechanotransduction persisted in the absence of Trpm2 (Fig 2A). To further test transduction, we
recorded hair cell transduction currents in response to a family of bundle deflections. Both
wild-type and Trpm2fl/fl:Gfi1-Cre+ outer hair cells showed high amplitude, rapidly adapting
transduction currents (Fig 2B), with peak currents in response to the largest deflections that
were not significantly different (Fig 2C).
The auditory brainstem response (ABR), a sound-evoked voltage change measured near the
brainstem, tests both transduction and synaptic function of the peripheral auditory system.
ABR responses to pure tone stimuli were largely normal in Trpm2-deleted animals (Fig 2D).
There was a slight elevation of hearing threshold at high frequencies in Trpm2-deficient mice,
but overall auditory function was preserved. Based on these results we conclude that Trpm2 is
not required for hair cell transduction.
Pkd2, Pkd2l1, Pkd2l2, and Pkd1l3 are not individually required for hair
cell transduction
Pkd2l1 also showed an intriguing expression pattern (Fig 1), with 6.3-fold enrichment in hair
cells compared to surrounding cells in both cochlea and utricle. The reported conductance of
Pkd2l1 channels, 120–200 pS [28,67], matched that expected for the transduction channel.
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
4 / 22
TRP Channels in Hair Cell Mechanotransduction
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
5 / 22
TRP Channels in Hair Cell Mechanotransduction
Fig 2. Trpm2 is not required for hair cell mechanotransduction. (a) FM1-43 accumulation by IHCs and OHCs
from control Trpm2fll+:Gfi1-Cre+ mice (top) and deleted Trpm2fl/fl:Gfi1-Cre+ mice (bottom). FM1-43 accumulation by
hair cells was similar in control and Trpm2-deleted hair cells. Age: P5+2div. Scale bar = 20 μm. (b) A family of OHC
transduction current recordings (top traces) in response to stereocilia bundle deflections (bottom traces) in WT (left)
and Trpm2fl/fl:Gfi1-Cre+ (right) mice. (c) Average peak transduction current (red traces in b) for control mice (WT and
Trpm2fl/+:Gfi1-Cre+) and Trpm2fl/fl:Gfi1-Cre+ mice. d, ABR thresholds in response to pure tone stimuli. Trpm2fl/fl:
Gfi1-Cre+ mice show normal hearing. Data are mean ± sem; n as indicated.
doi:10.1371/journal.pone.0155577.g002
Indeed, the PKD2 group of TRP channels has previously been suggested to include attractive
candidates for the transduction channel [21].
We obtained a Pkd2l1 knockout mouse lacking exons 3 and 4, which produced a premature
stop in exon 5 [77]. Using in situ hybridization in cochlear sections, an antisense probe showed
label in the hair-cell region (Fig 3A). No expression of Pkd2l1 mRNA was detected in Pkd2l1-/mice, confirming the knockout, or with sense probe, confirming the probe specificity. However
ABR measurements in Pkd2l1-/- mice showed normal hearing thresholds (Fig 3C), indicating
that mechanotransduction persists in the absence of Pkd2l1. In scanning electron microscopy
(SEM) images of Pkd2l1-/- mice at age P35, hair bundles appeared normal compared to wildtype controls (Fig 4A, 4C, 4E and 4G).
Fig 3. Neither Pkd2 nor Pkd2l1 is required for hair cell mechanotransduction. (a) In situ hybridization in cochlear sections. (Left) no label is evident
with a control sense probe. (Middle) no specific label is evident in the Pkd2l1 knockout. (Right) In situ hybridization with an antisense probe shows label of
inner hair cells (arrowhead), outer hair cells (arrows) and inner sulcus cells (asterisks) in the organ of Corti. Scale bar = 50 μm; age P2. (b) ABR thresholds
in response to pure tone stimuli. Pkd2l1-/- mice show normal hearing at age P31-P37. (c) Pkd2-/-:Atoh1-Cre+/-mice show normal hearing at age 4~6 weeks.
Data are mean ± sem; n as indicated.
doi:10.1371/journal.pone.0155577.g003
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
6 / 22
TRP Channels in Hair Cell Mechanotransduction
The PKD2 group of TRP channels is not very divergent (S1 Fig), and PKD2 channels interact with each other forming heteromultimeric channels, further indicating their similarity
[78,79,80,81]. These both raise the possibility that expression of Pkd2 or Pkd2l2 might compensate for the loss of Pkd2l1.
We therefore generated a conditional Pkd2 knock-out mouse line, by flanking exon 9
(aa630-671, comprising the pore and most of the sixth transmembrane domain) with loxP sites
Pkd2fl/fl; see Methods and S3 Fig). Hair-cell-specific Cre recombination was achieved by crossing Pkd2fl/fl mice with a mouse line expressing Cre recombinase under control of the Atoh1
enhancer, restricting the Cre expression to hair cells [82]. Hair cell-specific absence of Pkd2
expression in Atoh1-Cre mice was confirmed using inner ear genomic PCR (S3 Fig).
We first assessed the consequence of Pkd2 deletion alone. Pkd2fl/fl:Atoh1-Cre+ knockout
mice looked similar to their heterozygous control Pkd2fl/+:Atoh1-Cre+ littermates, suggesting
no gross developmental defects. Hair bundles in Pkd2 knockouts also appeared normal (Fig
4B–4F). ABR measurements showed normal hearing thresholds (Fig 3D), indicating that haircell mechanotransduction does not require Pkd2.
As an independent confirmation, we used a second Pkd2fl/fl mouse line [83] missing exons
11–13, and deleted Pkd2 in hair cells by crossing to Atoh1-Cre+. In heterozygous controls, an
antibody to Pkd2 labeled the kinocilia of hair cells and the primary cilia of adjacent supporting
cells (S4 Fig). In homozygous knockouts, antibody label was missing from kinocilia but not
adjacent primary cilia, confirming both the antibody specificity and the cell-specific deletion.
These Pkd2 knockouts have normal hearing as well (S4 Fig).
We then obtained Pkd2l2 knockout mice in which exons 3 and 4 were replaced with a LacZ
+Neo cassette, terminated by a stop (Jackson Laboratory B6.129P2- Pkd2l2tm1Dgen/J, Stock #:
005829). Because Pkd2l1 was thought to function together with Pkd1l3 in acid taste transduction [84,85,86], we also obtained a Pkd1l3 knockout line in which deletion of exons 17 through
21 produces a frame shift [87].
In double knockouts, we assessed stereocilia bundle morphology with SEM, and hearing
sensitivity with ABR. Double knockout mice with hair cells missing both Pkd2 and Pkd2l1
showed normal bundles (Fig 4D–4J) and had normal hearing thresholds (Fig 4M). Similarly,
single Pkd1l3 knockouts and double knockouts missing both Pkd2l1 and Pkd1l3 showed normal bundles (Fig 4H and 4I) and normal hearing thresholds (Fig 4N). Single Pkd2l2 knockouts
are also normal (Fig 4K–4O).
Triple Pkd2, Pkd2l1 and Pkd2l2 knockout mice show normal
transduction
Pkd2fl/fl:Atoh1-Cre+, Pkd2l1-/- and Pkd2l2-/- mouse lines were then crossed to each other to
generate triple knockout mice. Triple knockouts were viable and could be bred as homozygotes.
SEM imaging of stereocilia bundles revealed normal bundle morphology in adult mice (Fig
4L). To test transduction in cochlear hair cells missing Pkd2, Pkd2l1 and Pkd2l2, we first used
the FM1-43 dye accumulation assay in acutely dissected P6 organ of Corti epithelia. Triple
knockout mice showed similar levels of fluorescence dye intensity accumulation in hair cells as
compared to double knockout control mice, suggesting preserved transduction (Fig 5A). To
further test transduction, we recorded transduction currents in response to a family of bundle
deflections in triple knockout organ of Corti explants and in age-matched control wild-type
mice of the same background. Both wild-type and Pkd2fl/fl:Atoh1-Cre+:Pkd2l1-/-:Pkd2l2-/OHCs showed high amplitude, rapidly adapting transduction currents (Fig 5B). The peak
transduction current in response to the largest deflection was similar between the tested groups
(Fig 5C). Vestibular hair cells of Pkd2fl/fl:Atoh1-Cre+:Pkd2l1-/-:Pkd2l2-/- triple knockouts also
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
7 / 22
TRP Channels in Hair Cell Mechanotransduction
Fig 4. Mice lacking single or multiple PKD genes show normal stereocilia bundle morphology and hearing function. (a-d) SEM images of postnatal
4~6 weeks organ of Corti hair cells at low magnification in WT and PKD-deficient mice. Scale bar = 10 μm. (e-l) OHC bundles at high magnification. Scale
bar = 1 μm. (m) ABR thresholds in response to pure tone stimuli in Pkd2 and Pkd2l1 single and double knockouts. (n) Pkd2l1 and Pkd1l3 single and double
knockouts. (o) Pkd2l2 knockouts. No functional deficit was observed in any combination tested. Data shown as mean ± sem.
doi:10.1371/journal.pone.0155577.g004
showed transduction currents (data not shown). ABR responses to pure tone stimuli showed
similar threshold levels in Pkd2fl/fl:Atoh1-Cre+:Pkd2l1-/-:Pkd2l2-/- triple knockouts, as compared to their double knockout littermates with either Pkd2 or Pkd2l2 present (Fig 5D). Based
on these results we conclude that the PKD2 subfamily of channels is not required for hair cell
transduction.
Discussion
The TRP channel family is broadly expressed in a different cell types and activated by a variety
of stimuli, being especially prominent in reception of sensory stimuli. Like the hair-cell transduction channel, they are generally nonselective cation channels of high conductance. Some
TRPs are thought to sense mechanical stimuli [88,89], although activation may be indirect
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
8 / 22
TRP Channels in Hair Cell Mechanotransduction
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
9 / 22
TRP Channels in Hair Cell Mechanotransduction
Fig 5. Triple PKD knockout (Pkd2fl/fl:Atoh1-Cre+:Pkd2l1-/-:Pkd2l2-/-) mice show normal mechanotransduction and normal hearing in ABR
measurements. (a) FM1-43 accumulation is similar in IHCs and OHCs from P6 Pkd2fl/fl:Atoh1-Cre-:Pkd2l1-/-:Pkd2l2-/- controls lacking Cre (left) and
Pkd2fl/fl:Atoh1-Cre+:Pkd2l1-/-:Pkd2l2-/- knockout mice (right), suggesting normal transduction. Scale bar = 20 μm. (b) A family of OHC transduction
currents (top traces) in response to stereocilia bundle deflections (bottom traces) in WT (left) and Pkd2fl/fl:Atoh1-Cre+:Pkd2l1-/-:Pkd2l2-/- knockout (right)
mice. (c) Average peak transduction currents (red traces in b) for WT mice (blue; n = 5) and triple knockout (red bar, n = 6). (d) ABR threshold
measurements in response to pure tone stimuli in Pkd2fl/fl:Atoh1-Cre+:Pkd2l1-/-:Pkd2l2-/- knockout mice indicate normal hearing. Data are mean ± sem.
doi:10.1371/journal.pone.0155577.g005
[90]. This has led to the consideration of various TRPs as components of the hair cell mechanotransduction channel. Here we evaluate each of the six branches of the TRP family in mice.
TRPMLs
The varitint-waddler mutant (Va(J)) of Trpml3 (mucolipin 3; Mcoln3) has profound hearing
loss [91] and impaired transduction in hair cells [64,92]. However, it was found that the Va(J)
mutation leads to constitutive activation of the channel, causing a continuous inward current
and consequent cell death. Hearing loss is explained by the loss of hair cells [20,61,63,64,92].
Hearing is normal in a null mutant of Trpml3 [93].
The TRPML branch is not very divergent (S1 Fig) so there is the possibility of compensation
for loss of Trpml3 by a related gene. Trpml2 has negligible expression in the inner ear, but
Trpml1 is expressed in both hair cells and surrounding cells (Fig 1). However the channel conductance for this branch is not consistent with the transduction channel conductance, and
Trpml1 protein is mainly located in late endosomes and lysosomes [94].
PKDs
An analysis of candidates for the hair cell transduction channel pointed out that some members of the PKD (TRPP) branch of the TRP family have properties similar to those of the transduction channel [21]. PKD2 channels are highly permeable to calcium: Pkd2 has a PCa/PNa of 5
whereas Pkd2l1 has a PCa/PNa of ~4 [95]. The portfolio of blocking ions is similar to that of
hair cells: Pkd2 is blocked by Ca2+, La3+ and Gd3+, and Pkd2l1 is blocked by Ca2+, Mg2+, La3+
and Gd3+. Both Pkd2 and Pkd2l1 are blocked by amiloride (IC50 = 40–130 μm). The conductance of Pkd2 is 40–170 pS [40,48], and Pkd2l1 conductance is ~120 pS for inward current
[67,96]. Pkd2, Pkd2l1 and Pkd2l2 are all expressed in hair cells, with the Pkd2l1 profile being
especially appropriate [25]. Moreover, some studies have suggested that the Pkd2 channel is
mechanosensitive—specifically that it is located in primary cilia, where it is activated by cilia
bending from environmental fluid flow [97,98,99].
Because constitutive deletion of the Pkd2 gene is embryonic lethal, we used an Atoh1-Cre
line to delete Pkd2 conditionally in hair cells. Using a pure-tone ABR test, we found that hearing is normal in Pkd2 conditional knockout mice (Fig 3D). SEM imaging showed a normal hair
bundle in the cochlea. Testing a second, independent Pkd2 conditional knockout line which
truncates the C-terminal of Pkd2 channels, we found similarly normal function. Furthermore,
antibody labeling showed that in hair cells Pkd2 is located in the kinocilia but not stereocilia.
Finally, we recently re-analyzed mechanosensitivity in primary cilia in a variety of cell types
and found that none shows rapid influx of Ca2+ upon cilium bending, casting doubt on the
idea that Pkd2 is a mechanically gated channel [67,96,100].
We also investigated if Pkd2l1 is involved in hair cell transduction, using a Pkd2l1 knockout
mouse that we recently described [77]. RT-PCR from whole mouse cochlea showed that
expression of Pkd2l1 is correlated with the acquisition of mechanotransduction [24], and
RNA-seq showed it is specifically expressed in hair cells at the start of mechanosensitivity (Fig
1). A sensitive mass spectroscopy study found that Pkd2l1 protein is enriched in hair bundles
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
10 / 22
TRP Channels in Hair Cell Mechanotransduction
of the chicken auditory epithelium [101]. In situ hybridization in mouse showed that Pkd2l1 is
highly expressed in hair cells and also some inner sulcus cells (Fig 3A). However the Pkd2l1
knockout mouse did not show abnormalities in hearing, using the ABR test (Fig 3C), or abnormal hair bundle morphology (Fig 4).
Pkd1l3 had been thought to form a heteromeric complex with Pkd2l1 for sour taste sensation [85,102,103], suggesting that it might share function with Pkd2l1 in hair cells. Pkd1l3 was
also found to be expressed in the auditory system [24], although expression in hair cells is limited (Fig 1). However we found that a single knockout of Pkd1l3 and a double knockout with
Pkd2l1 showed no abnormalities in the ABR or hair bundle morphology (Fig 4).
Pkd2l2 is also expressed in hair cells, although not preferentially (Fig 1). We found that a
single Pkd2l2 knockout has a normal hearing sensitivity and hair bundle morphology. Because
of the possibility of compensation among this closely-related group, we created double and triple knockouts of Pkd2, Pkd2l1 and Pdk2l2. Even triple knockouts showed no deficit in hearing
sensitivity, bundle morphology, or mechanotransduction assessed with FM1-43 accumulation
and single-cell recording. We can thus rule out the PKD2 branch of the TRP family.
In addition to Pkd1l3, some of the Pkd1 group of TRPs are sparsely expressed in hair cells.
However none of the Pkd1 group have robust and hair-cell-specific expression profiles (Fig 1).
Pkd1 mutants have normal transduction current and only moderate loss of hearing sensitivity
[104]. Moreover, PKD1s are thought to form ion channels as heteromultimers with PKD2
channels [105], and the PKD2s have been ruled out.
The entire PKD group of TRP channels is thus unlikely to participate in hair-cell
transduction.
TRPA
Trpa1 was also considered as a strong transduction channel candidate because knockdown of
expression in both mice (with virally delivered siRNA) and zebrafish (morpholino injection)
reduced the transduction current, and because it has a large single-channel conductance of 250
pS in low Ca2+ [106], (data not shown). However, mice lacking Trpa1 have normal transduction current and hearing [70]. Trpa1 is the only member of the Trpa branch in mice, so compensation by a related gene is unlikely.
TRPCs
Although Trpc1 is expressed in mouse hair cells (Fig 1), the other members of this branch do
not have an appropriate expression pattern and Trpc4 is not expressed at all. In addition, a quadruple knockout of Trpc1, Trpc3, Trpc5 and Trpc6 shows normal hearing sensitivity and hair
cell morphology [71,72]. We previously showed that the remaining TRPC gene, Trpc2, is highly
expressed in mouse vomeronasal neurons and the protein is located in the sensory cilia where
it is thought to participate in pheromone transduction [107]. Trpc2 knockout mice do lack
pheromone sensitivity but have no vestibular deficit [108]. Also, TRPC2 is a nonfunctional
pseudogene in a species (human) that has normal hearing [109]. Finally, the single channel
conductance of the TRPC channels is lower than that of the hair cell transduction channel.
TRPMs
In situ hybridization and RNA-seq showed Trpm2 to be selectively expressed by inner ear hair
cells, suggesting that it plays an important role in these cells [25]. We found, however, that
Trpm2 knockout mice exhibited normal transduction current and FM-143 accumulation.
Trpm1 knockouts also have normal hearing [73]. It might be that other TRPMs compensate in
these knockouts, but—with the exception of Trpm7—none of the others has an appropriate
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
11 / 22
TRP Channels in Hair Cell Mechanotransduction
expression pattern or single-channel conductance. Trpm7 is expressed but not enriched in hair
cells, its single channel conductance is high but not as high as the transduction channel, and
zebrafish studies suggest it is not required for hair cell transduction in zebrafish [110]. Thus
none of the TRPMs seems likely to be a transduction channel.
TRPVs
Trpv4 was first suggested as a possible component of a heteromeric transduction channel
because it was mechanosensitive—responding to changes in osmolarity—and was expressed in
auditory hair cells [111]. Newer methods have shown the Trpv4 gene to be expressed only at
very low levels in cochlear hair cells and not at all in utricular hair cells [25]. Although Trpv4
knockout mice show mild, late onset hearing loss, there is no deficit in hearing at earlier ages,
excluding it as a transduction channel candidate [72].
Of other TRPVs, only Trpv3 has a large conductance; however its expression pattern is not
consistent with the hair cell transduction channel (Fig 1). None of the other TRPV channels
shows an appropriate expression pattern in hair cells.
Conclusion
Although the TRP family of ion channels initially seemed like a rich source of candidates for
the hair-cell transduction channel, most of them can be ruled out by inappropriate expression
pattern, inappropriate conductance, or lack of an auditory or vestibular phenotype in knockout
mice. Here, we have tested the remaining reasonable candidates—including some that were
quite attractive based on expression and conductance—but found no deficit in transduction in
hair cells of these animals. Although many TRPs are expressed in hair cells and many are sure
to carry out important functions in these cells, it now seems safe to exclude the entire TRP family in the search for a transduction channel.
Materials and Methods
TRP channel expression data
Expression data for all 33 mouse TRP channels were drawn from Scheffer et al., 2015, as posted
on the SHIELD database (shield.hms.harvard.edu). Read counts were analyzed based on the
20207 RefSeq genes in the DNAnexus set. Pkd1l1 is not in that set, so reads for Pkd1l1 were
identified by specifically querying the raw data set.
Mouse lines
This study was carried out in strict accordance with the recommendations in the Guide for the
Care and Use of Laboratory Animals of the National Institutes of Health. The protocol was
approved by the Institutional Animal Care and Use Committee at Harvard Medical School
(Protocol Number: 03524). ABR measurements were performed under anesthesia (ketamine
(100 mg/kg)/xylazine (10 mg/kg) cocktail). Newborn mice (P0-P4) were anesthetized by cooling, then euthanized by decapitation. Adult mice were euthanized by isoflurane overdose, followed by cervical dislocation. All efforts were made to minimize suffering. All primers used in
current study are summarized in Table 1.
Trpm2 conditional knockout mouse line. We worked with Ingenious Targeting Laboratory (Ronkonkoma, New York, USA) to make the Trpm2 conditional knockout. A 10.2-kb
Trpm2 genomic fragment was subcloned from a C57BL/6 BAC clone to construct the targeting
vector using homologous recombination. A FRT-LoxP-Neo-FRT-LoxP cassette was inserted
upstream of exon 21 and the third single LoxP site was inserted downstream of exon 21 (S2
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
12 / 22
TRP Channels in Hair Cell Mechanotransduction
Table 1. Primers used for genotyping and validation of gene deletion.
Mouse lines
Primer Name
PCR primer
concentration
Sequence
Floxed
allele
WT
allele
Trpm2cKO
(David Corey)
LOX
1.2 μM
TGAGGCGGAAGGAATTAGCAC
320
259
Pkd2cKO (Jing
Zhou)
Pkd2cKO (Terry
Watnick)
Pkd2l1
Gfi1-Cre
Atoh1-Cre
Pkd2l2
Pkd1l3
KO
allele
Purpose
Genotyping
SDL
1.2 μM
CCCACCTGACAGTCACAAGTGTG
320
259
TM2cKO15101f
1.2 μM
GACTTCATCATGTTCTGTCT
4184 (N/
A*)
2415
(N/A*)
mPKD2in9F3
0.6 μM
TTGTGCATTTGGTGATGTGTTA
520
468
mPKD2in9R3
0.6 μM
CCACATTTACATGGCATCTGAG
520
468
PKD2-5940f1
1.2 μM
AAGCTGTGTTATCATTCTAGAAAGC
>2457
2457
MG/3 flox c-f
1.2 μM
GGGGTTTCCTATGAAGAGTTCCAAG
485
396
MG/3 flox d-r
1.2 μM
CTGACAGGCACCTACAGAACAGTG
485
Neo82
0.6 μM
CTGCCTTGGGAAAAGCGCCT
~520
~480
Genotyping
MPclF904
0.6 μM
AAGATCAGCTCCCCTTTGGACCT
~520
~480
Genotyping
R520
0.6 μM
TTCCACCCCAGGATTCTCTG
~520
~480
Genotyping
Genotyping
Genotyping
616
Verification cKO in
the inner ear
Genotyping
Genotyping
344
Verification cKO in
the inner ear
Genotyping
396
Genotyping
Gfi-1F
1.2 μM
GGG ATA ACG GAC CAG TTG
609
672
Gfi1Cre-R
1.2 μM
GCC CAA ATG TTG CTG GAT AGT
609
672
Genotyping
Gfi-1R
0.6 μM
CCG AGG GGC GTT AGG ATA
609
672
Genotyping
Atoh1Cre-F
1.2 μM
ATCGGCCTCCTCCTCGTAGACAGC
550
Genotyping
Atoh1Cre-R
1.2 μM
GGATCCGCCGCATAACCAGTGA
550
Genotyping
IMR4245
0.3 μM
CATCATCAGGTAGAGAAGTGTCCAC
~200
~450
Genotyping
IMR4246
1.2 μM
CGTGCGTGCAAACACCCACACACAG
~200
~450
Genotyping
IMR5100
1.2 μM
TTCAACAGACCTTGCATTCCTTTGG
~200
~450
Genotyping
1111nF-5’
0.3 μM
AGGAGAGGATTGACTTCTATGAG
~250
650
Genotyping
1112nR-5’
0.3 μM
CAGGAGAGCCTCTGGACTCGTGT
~250
650
Genotyping
1328F-5’
1.2 μM
GATGGAAGCCGGTCTTGTCGAT
~250
650
Genotyping
1538R-5’
1.2 μM
TCGAGCCCCAGCTGGTTCTTTCC
~250
650
Genotyping
* Not amplified
doi:10.1371/journal.pone.0155577.t001
Fig). The region flanked by the second and the third LoxP sites is about 2.0 kb. Exon 21
encodes amino acids 929–985 and corresponds to the fifth transmembrane domain and pore of
Trpm2; its deletion also causes an early stop. A homology short arm extends 1.8 kb to the 3’ of
the FRT-LoxP-Neo-FRT-LoxP cassette whereas a homology long arm extends 6.4 kb from the
5’ side of the third LoxP site. The linearized targeting vector was then electroporated into iTL
BA1(C57BL/6X129/SvEv) hybrid embryonic stem cells.
Pkd2l1 constitutive knockout. We have previously described the Pkd2l1 knockout [77]. It
lacks exons 3 and 4, leading to a premature stop codon in exon 5.
Pkd2 conditional knockout. A 7.3kb genomic fragment of the Pkd2 gene was used to construct the targeting vector. Three LoxP sites were inserted as shown in S3 Fig. The first LoxP
site was inserted upstream of exon 9 and a LoxP-Neo-LoxP cassette was inserted downstream
of exon 9. The first and second LoxP sites thus flank a 2.2kb genomic region that includes exon
9. Exon 9 encodes amino acids 630–671, comprising the pore and most of the sixth transmembrane domain of Pkd2. The homology short arm extended 0.9 kb to the first loxP site and a
homology long arm extended 4.2 kb downstream of the LoxP-Neo-LoxP cassette.
Pkd2l2 constitutive knockout. Mice were obtained from The Jackson Laboratory
(B6.129P2-Pkd2l2tm1Dgen/J, #005829). In this mutant mouse line, a lacZ-Neo cassette was
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
13 / 22
TRP Channels in Hair Cell Mechanotransduction
inserted into the Pkd2l2 gene to replace a 1.8-kb genomic fragment including exons 3 and 4. As
a result, the endogenous Pkd2l2 gene promoter drives expression of the beta-galactosidase gene
but not the endogenous Pkd2l2 gene (S5 Fig).
Pkd1l3 constitutive knockout. Mice were obtained from The Jackson Laboratory
(B6;129S4-Pkd1l3tm1Sul/J, Stock #: 008419). They lack exons 17 through 21, which encode
transmembrane domains 2–5 [87].
Cre lines. The Gfi1-Cre mouse line was generously provided by Dr. Lin Gan (University of
Rochester)[75]. The Atoh1-Cre mouse was obtained from The Jackson Laboratory (B6.Cg-Tg
(Atoh1-cre)1Bfri/J, #011104) [82].
Immunofluorescence
Mouse cochleas and/or utricles were dissected from P2~P8 mutant mice and their wild-type littermates. Samples were fixed with 4% formaldehyde for 2 hr, rinsed with PBS, incubated in 0.1
M citrate buffer (pH 7.0) at 60°C for 30 min for antigen retrieval. Whole mount samples were
permeabilized with 0.5% Triton X-100 for 30 min, and blocked with 10% goat serum supplemented with 0.5% Triton X-100 for 30 min. Samples were then incubated with primary antibody overnight at 4°C, rinsed with PBS, and further incubated in secondary antibodies
together with phalloidin for 6 hr. Samples were then mounted with Prolong Gold antifade kit
(Invitrogen), cured in the dark at room temperature overnight and imaged with an upright
Olympus FluoView FV1000 confocal laser scanning microscope (60X 1.42NA objective).
ABR measurements
The ABR assay was performed using a Tucker Davis Technologies (TDT, Gainsville, FL) workstation (System III). Mice age P28 to P60 were anesthetized by intraperitoneal injection of a ketamine (100 mg/kg)/xylazine (10 mg/kg) cocktail. Anesthetized mice were then placed on a heating
pad and electrodes were placed subcutaneously in the vertex, underneath the left ear, and on the
back near the tail. Tone stimuli of 4, 5.6, 8, 11.2, 16, 22, 32 and 45.3 kHz were calibrated with a
precision microphone system (PS9200Kit, ACO Pacific, USA) using the TDT SigCal software
package. The recorded signals were band-pass filtered (300 Hz to 3 kHz) and amplified 100,000
times. The number of acquisition trials was set to 500 averages. Maximum stimulus intensity was
set to 95 dB peak SPL with attenuation decreasing from 85 dB to 0 dB SPL at 5 dB SPL intervals.
All ABR thresholds were read by second investigator who was blind to the mouse genotype.
Field emission scanning electron microscopy
Cochleas from either P2 or adult (P28-P40) mice were dissected out and immediately
immersed in 0.5% glutaraldehyde / 0.1 M sodium cacodylate buffer / 3 mM CaCl2 (pH 7.3) for
2 hr. Adult cochleas were further decalcified in 120 mM EDTA (pH 7.2) for 24 hr. Cochlea
coils were dissected out in distilled water. Samples were processed through an OTOTO procedure [112] with modifications. Briefly, samples were washed in the cacodylate dilution buffer
three times for 5 min each, fixed in 1% OsO4 in cacodylate buffer for 1 hr, and washed in H2O
for 5 min three times. The samples then went through 1% freshly prepared tannic acid for 1 hr,
then 1% OsO4 for 1 hr, 1% tannic acid for 1 hr, and 1% OsO4 for 1 hr, with an H2O wash
between steps. Next, samples were washed and processed through the crescent isopropanol
series steps (30% isopropanol at room temperature, 50% isopropanol on ice, 70% isopropanol
at -20°C, 90% isopropanol slurry on dry ice, 95% isopropanol slurry on dry ice, 100% isopropanol slurry on dry ice), with each step exposition set to 15 min. Sample were then critical point
dried, sputter coated with platinum and imaged on a Hitachi S-4800 field emission scanning
electron microscope.
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
14 / 22
TRP Channels in Hair Cell Mechanotransduction
Whole-cell patch clamp recording
Organ of Corti epithelia were dissected at P3-P5 in L-15 medium (Life Technologies), placed in
glass bottom Petri dishes (WPI Inc.) and cultured for 2–3 days in DMEM/F12 medium (Life
Technologies) supplemented with 5% FBS and 10 mg/l ampicillin at 37°C (10% CO2) as previously described [113]. Experiments were performed at room temperature in L-15 containing
the following inorganic salts (in mM): NaCl (137), KCl (5.4), CaCl2 (1.26), MgCl2 (1.0),
Na2HPO4 (1.0), KH2PO4 (0.44), MgSO4 (0.81). Hair cells were observed with an inverted
microscope (TE 2000, Nikon) using a 100X 1.3 NA oil-immersion objective lens and DIC
optics. Pipettes for whole-cell patch-clamp recordings were filled with intracellular solution
containing (in mM): CsCl (140), MgCl2 (2.5), Na2ATP (2.5), EGTA (1.0), HEPES (5.0). The
pipette resistance was typically 4–6 MΩ when measured in the bath. Patch clamp recordings
were performed with an AxoPatch 200B amplifier (Molecular Devices) controlled by pClamp 9
software package. Hair cells were held at –60 mV between the short periods of transduction
recordings, when the holding potential was temporarily hyperpolarized to –90 mV. All
recorded hair cells were located 35–45% away from the apex of the organ of Corti explant.
Hair bundles were deflected using a stiff glass probe, fire-polished to fit the shape of the
stereocilia bundle (~5–7 μm). The probe was mounted on a piezo actuator (PA 8/14 SG, Piezosystem Jena), equipped with a strain gauge sensor to provide a direct reading of the probe’s
axial displacement. The piezo was driven by a custom-made amplifier, providing a rapid step
deflection within ~40 μs (10–90% risetime). The angle between the axis of the probe movement
and the bottom surface of a dish was kept constant at ~30 degrees.
FM1-43 loading
Organ of Corti epithelia were acutely dissected from P4-P5 mice in L-15 cell culture medium,
and either mounted on coverslips using tungsten minutien pins (WPI Inc.) for the experiment,
or cultured for an additional 2 days as described above. Following tectorial membrane removal
and medium aspiration, FM1-43 solution (2 μM in L-15) was applied to the tissue for 30–60 s,
then quickly aspirated; the tissue was rinsed once with L-15 and the excessive dye quenched by
a 0.2 mM solution of 4-sulphonato calix[8]arene, sodium salt (SCAS, Biotium) in L-15. The
organ of Corti was then observed on an upright Olympus FV1000 confocal microscope,
equipped with 60X 1.1 NA water-dipping objective lens.
Supporting Information
S1 Fig. Mouse TRP channel phylogeny. There are 33 genes in six major groups (TRPM,
TRPC, TRPV, PKD/TRPP, TRPML and TRPA). Length of line indicates divergence.
(TIF)
S2 Fig. Strategy and validation of Trpm2 knockout. (a) Trpm2 conditional knockout strategy.
We inserted two LoxP sites to flank a ~1.9 kb region that includes exon 21. A LoxP-FRT-NeoLoxP-FRT cassette was inserted upstream of exon 21 and the third LoxP site was inserted
downstream of exon 21. Exon 21 encodes aa929-985 including the essential fifth transmembrane domain and pore. The deletion also led to a downstream frameshift. Red arrowheads
refer to LoxP sites, yellow brackets refer to FRT sites, and green arrows refer to the neomycin
resistance gene. Primers used for genotyping and validation are indicated as gray arrows. (b)
PCR from genomic DNA purified from inner ears of Trpm2fl/fl: Gfi1-Cre+ mice, Trpm2fl/fl:
Gfi1-Cre- mice, and an age-matched wildtype mouse. (Left) PCR results using primer pair
TM2cKO15101f and SDL. Lane 1 (Trpm2fl/fl:Gfi1-Cre+) shows a fused short band (616bp) only
from the deleted allele; lane 2 lacking Cre (Trpm2fl/fl:Gfi1-Cre-) and Lane 3 (wildtype) do not
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
15 / 22
TRP Channels in Hair Cell Mechanotransduction
show it. The fused band was confirmed by Sanger sequencing. (Middle) PCR bands at 320 bp
(floxed allele) in lane 1 and lane 2, and 259 bp (Wt allele)in lane 3, using primers LOX and
SDL, (Right) Genotyping for the Gfi1-Cre allele. A 1-kb DNA extension ladder was used (5 μl;
Invitrogen #10511–012).
(TIF)
S3 Fig. Strategy and validation of Pkd2 knockout. (a) Exon 9 of the Pkd2 gene was deleted by
flanking the ~2.2 kb targeting region with two LoxP sites. A LoxP-Neo-LoxP cassette was
inserted downstream of exon 9 and the third LoxP site was inserted upstream. Exon 9 encodes
aa630-671 including the pore domain and most of the sixth transmembrane domain of the
Pkd2 channel. Primers used for genotyping and validation are indicated as a gray arrows. (b)
PCR from genomic DNA purified from inner ears of Pkd2fl/fl:Atoh1-Cre+ mice, Pkd2 fl/fl:
Atoh1-Cre-/- mice, and an age-matched wildtype mouse. (Left) PCR results using primer pair
PKD2-5940f1 and mPKD2in9R3. Lane 1 (Pkd2fl/fl:Atoh1-Cre+) shows a fused short band (344
bp) from the deleted allele and a longer band (>2437 bp) from the genomic DNA cells without
Cre activity. The short fused band was confirmed by Sanger sequencing. Lane 2 (Pkd2 fl/fl:
Atoh1-Cre-) shows the same long band as in lane 1 but no short band. Lane 3 shows a band of
2457 bp produced from a wildtype mouse inner ear. (Middle) PCR produced bands at 520bp in
lane 1 and lane 2, and 468 bp in lane 3 using mPKD2in9F3 and mPKD2inR3. (Right) Genotyping of the Atoh1-Cre allele. A 1-kb DNA extension ladder was used (5 μl; Invitrogen #10511–
012).
(TIF)
S4 Fig. Pkd2 localization and Pkd2 knockout ABR data, in the second Pkd2 knockout
mouse [83]. (a,a’) Antibody labeling (Santa Cruz Biotechnology, #sc-10376) for Pkd2 (red)
and phalloidin staining for actin (green) in the heterozygote (Pkd2fl/+: Atoh1-Cre+) positivecontrol cochlea. Pkd2 label is evident in hair-cell kinocilia (arrowheads) and some supporting
cell primary cilia (arrows). (b,b’) Pkd2fl/fl: Atoh1-Cre+ knockout cochlea. Pkd2 label is absent
from hair cell kinocilia but not from supporting cell cilia. (c-c’) In vestibular hair cells of heterozygote mice, Pkd2 label is also in kinocilia. (d) ABR thresholds in response to pure tone stimuli. Pkd2fl/fl: Atoh1-Cre+ knockout mice show normal hearing. Data are mean ± SEM.
(TIF)
S5 Fig. Pkd2l2 knockout. The generation of the Pkd2l2 knockout mouse, based on information from The Jackson Laboratory (Pkd2l2tm1Dgen/J; stock #005829; https://www.jax.org/strain/
005829). A bacterial lacZ gene fused with a neomycin resistance gene replaced ~ 1.8 kb genomic sequence extending from the 3’ part of exon 3 to the 5’ part of exon 4. Thus the endogenous promoter drove the expression of beta-galactosidase.
(TIF)
Acknowledgments
We thank William Fowle (Northeastern University), and Jun Shen, Hongyu Zhao, Bruce Derfler and Yaqiao Li (Harvard Medical School) for advice and assistance.
Author Contributions
Conceived and designed the experiments: XW AAI PDN RMW MAV DPC. Performed the
experiments: XW AAI PDN RMW. Analyzed the data: XW AAI PDN RMW DPC. Contributed reagents/materials/analysis tools: MG-G TW JZ. Wrote the paper: XW AAI DPC.
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
16 / 22
TRP Channels in Hair Cell Mechanotransduction
References
1.
Hudspeth AJ, Corey DP (1977) Sensitivity, polarity, and conductance change in the response of vertebrate hair cells to controlled mechanical stimuli. Proc Natl Acad Sci U S A 74: 2407–2411. PMID:
329282
2.
Corey DP, Hudspeth AJ (1979) Response latency of vertebrate hair cells. Biophys J 26: 499–506.
PMID: 318064
3.
Corey DP, Hudspeth AJ (1979) Ionic basis of the receptor potential in a vertebrate hair cell. Nature
281: 675–677. PMID: 45121
4.
Ohmori H (1985) Mechano-electrical transduction currents in isolated vestibular hair cells of the chick.
J Physiol 359: 189–217. PMID: 2582113
5.
Lumpkin EA, Marquis RE, Hudspeth AJ (1997) The selectivity of the hair cell's mechanoelectricaltransduction channel promotes Ca2+ flux at low Ca2+ concentrations. Proc Natl Acad Sci U S A 94:
10997–11002. PMID: 9380748
6.
Crawford AC, Evans MG, Fettiplace R (1991) The actions of calcium on the mechano-electrical transducer current of turtle hair cells. J Physiol 434: 369–398. PMID: 1708822
7.
Kimitsuki T, Nakagawa T, Hisashi K, Komune S, Komiyama S (1996) Gadolinium blocks mechanoelectric transducer current in chick cochlear hair cells. Hear Res 101: 75–80. PMID: 8951434
8.
Farris HE, LeBlanc CL, Goswami J, Ricci AJ (2004) Probing the pore of the auditory hair cell mechanotransducer channel in turtle. J Physiol 558: 769–792. PMID: 15181168
9.
Geleoc GS, Lennan GW, Richardson GP, Kros CJ (1997) A quantitative comparison of mechanoelectrical transduction in vestibular and auditory hair cells of neonatal mice. Proc Biol Sci 264: 611–621.
PMID: 9149428
10.
Ricci AJ, Crawford AC, Fettiplace R (2003) Tonotopic variation in the conductance of the hair cell
mechanotransducer channel. Neuron 40: 983–990. PMID: 14659096
11.
Beurg M, Evans MG, Hackney CM, Fettiplace R (2006) A large-conductance calcium-selective
mechanotransducer channel in mammalian cochlear hair cells. J Neurosci 26: 10992–11000. PMID:
17065441
12.
Pan B, Geleoc GS, Asai Y, Horwitz GC, Kurima K, Ishikawa K, et al. (2013) TMC1 and TMC2 are components of the mechanotransduction channel in hair cells of the mammalian inner ear. Neuron 79:
504–515. doi: 10.1016/j.neuron.2013.06.019 PMID: 23871232
13.
Rusch A, Kros CJ, Richardson GP (1994) Block by amiloride and its derivatives of mechano-electrical
transduction in outer hair cells of mouse cochlear cultures. J Physiol 474: 75–86. PMID: 7516972
14.
Gale JE, Marcotti W, Kennedy HJ, Kros CJ, Richardson GP (2001) FM1-43 dye behaves as a permeant blocker of the hair-cell mechanotransducer channel. J Neurosci 21: 7013–7025. PMID:
11549711
15.
Kroese AB, Das A, Hudspeth AJ (1989) Blockage of the transduction channels of hair cells in the bullfrog's sacculus by aminoglycoside antibiotics. Hear Res 37: 203–217. PMID: 2468634
16.
Ricci A (2002) Differences in mechano-transducer channel kinetics underlie tonotopic distribution of
fast adaptation in auditory hair cells. J Neurophysiol 87: 1738–1748. PMID: 11929895
17.
Marcotti W, van Netten SM, Kros CJ (2005) The aminoglycoside antibiotic dihydrostreptomycin rapidly
enters mouse outer hair cells through the mechano-electrical transducer channels. J Physiol 567:
505–521. PMID: 15994187
18.
Pan B, Waguespack J, Schnee ME, LeBlanc C, Ricci AJ (2012) Permeation properties of the hair cell
mechanotransducer channel provide insight into its molecular structure. J Neurophysiol 107: 2408–
2420. doi: 10.1152/jn.01178.2011 PMID: 22323630
19.
Vollrath MA, Kwan KY, Corey DP (2007) The micromachinery of mechanotransduction in hair cells.
Annu Rev Neurosci 30: 339–365. PMID: 17428178
20.
Cuajungco MP, Grimm C, Heller S (2007) TRP channels as candidates for hearing and balance
abnormalities in vertebrates. Biochim Biophys Acta 1772: 1022–1027. PMID: 17300924
21.
Fettiplace R (2009) Defining features of the hair cell mechanoelectrical transducer channel. Pflugers
Arch 458: 1115–1123. doi: 10.1007/s00424-009-0683-x PMID: 19475417
22.
Geleoc GS, Holt JR (2003) Developmental acquisition of sensory transduction in hair cells of the
mouse inner ear. Nat Neurosci 6: 1019–1020. PMID: 12973354
23.
Lelli A, Asai Y, Forge A, Holt JR, Geleoc GS (2009) Tonotopic gradient in the developmental acquisition of sensory transduction in outer hair cells of the mouse cochlea. J Neurophysiol 101: 2961–2973.
doi: 10.1152/jn.00136.2009 PMID: 19339464
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
17 / 22
TRP Channels in Hair Cell Mechanotransduction
24.
Asai Y, Holt JR, Geleoc GS (2010) A quantitative analysis of the spatiotemporal pattern of transient
receptor potential gene expression in the developing mouse cochlea. J Assoc Res Otolaryngol 11:
27–37. doi: 10.1007/s10162-009-0193-8 PMID: 19834762
25.
Scheffer DI, Shen J, Corey DP, Chen ZY (2015) Gene Expression by Mouse Inner Ear Hair Cells during Development. J Neurosci 35: 6366–6380. doi: 10.1523/JNEUROSCI.5126-14.2015 PMID:
25904789
26.
Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, Julius D (1997) The capsaicin
receptor: a heat-activated ion channel in the pain pathway. Nature 389: 816–824. PMID: 9349813
27.
Zitt C, Obukhov AG, Strubing C, Zobel A, Kalkbrenner F, Luckhoff A, et al. (1997) Expression of
TRPC3 in Chinese hamster ovary cells results in calcium-activated cation currents not related to store
depletion. J Cell Biol 138: 1333–1341. PMID: 9298988
28.
Chen XZ, Vassilev PM, Basora N, Peng JB, Nomura H, Segal Y, et al. (1999) Polycystin-L is a calcium-regulated cation channel permeable to calcium ions. Nature 401: 383–386. PMID: 10517637
29.
Hofmann T, Obukhov AG, Schaefer M, Harteneck C, Gudermann T, Schultz G (1999) Direct activation
of human TRPC6 and TRPC3 channels by diacylglycerol. Nature 397: 259–263. PMID: 9930701
30.
Nilius B, Vennekens R, Prenen J, Hoenderop JG, Bindels RJ, Droogmans G (2000) Whole-cell and
single channel monovalent cation currents through the novel rabbit epithelial Ca2+ channel ECaC. J
Physiol 527 Pt 2: 239–248. PMID: 10970426
31.
Strotmann R, Harteneck C, Nunnenmacher K, Schultz G, Plant TD (2000) OTRPC4, a nonselective
cation channel that confers sensitivity to extracellular osmolarity. Nat Cell Biol 2: 695–702. PMID:
11025659
32.
Vennekens R, Hoenderop JG, Prenen J, Stuiver M, Willems PH, Droogmans G, et al. (2000) Permeation and gating properties of the novel epithelial Ca(2+) channel. J Biol Chem 275: 3963–3969.
PMID: 10660551
33.
Nadler MJ, Hermosura MC, Inabe K, Perraud AL, Zhu Q, Stokes AJ, et al. (2001) LTRPC7 is a Mg.
ATP-regulated divalent cation channel required for cell viability. Nature 411: 590–595. PMID:
11385574
34.
Perraud AL, Fleig A, Dunn CA, Bagley LA, Launay P, Schmitz C, et al. (2001) ADP-ribose gating of
the calcium-permeable LTRPC2 channel revealed by Nudix motif homology. Nature 411: 595–599.
PMID: 11385575
35.
Runnels LW, Yue L, Clapham DE (2001) TRP-PLIK, a bifunctional protein with kinase and ion channel
activities. Science 291: 1043–1047. PMID: 11161216
36.
Sano Y, Inamura K, Miyake A, Mochizuki S, Yokoi H, Matsushime H, et al. (2001) Immunocyte Ca2+
influx system mediated by LTRPC2. Science 293: 1327–1330. PMID: 11509734
37.
Strubing C, Krapivinsky G, Krapivinsky L, Clapham DE (2001) TRPC1 and TRPC5 form a novel cation
channel in mammalian brain. Neuron 29: 645–655. PMID: 11301024
38.
Voets T, Prenen J, Fleig A, Vennekens R, Watanabe H, Hoenderop JG, et al. (2001) CaT1 and the
calcium release-activated calcium channel manifest distinct pore properties. J Biol Chem 276:
47767–47770. PMID: 11687570
39.
Yue L, Peng JB, Hediger MA, Clapham DE (2001) CaT1 manifests the pore properties of the calciumrelease-activated calcium channel. Nature 410: 705–709. PMID: 11287959
40.
Gonzalez-Perrett S, Batelli M, Kim K, Essafi M, Timpanaro G, Moltabetti N, et al. (2002) Voltage
dependence and pH regulation of human polycystin-2-mediated cation channel activity. J Biol Chem
277: 24959–24966. PMID: 11991947
41.
LaPlante JM, Falardeau J, Sun M, Kanazirska M, Brown EM, Slaugenhaupt SA, et al. (2002) Identification and characterization of the single channel function of human mucolipin-1 implicated in mucolipidosis type IV, a disorder affecting the lysosomal pathway. FEBS Lett 532: 183–187. PMID: 12459486
42.
Voets T, Prenen J, Vriens J, Watanabe H, Janssens A, Wissenbach U, et al. (2002) Molecular determinants of permeation through the cation channel TRPV4. J Biol Chem 277: 33704–33710. PMID:
12093812
43.
Watanabe H, Vriens J, Suh SH, Benham CD, Droogmans G, Nilius B (2002) Heat-evoked activation
of TRPV4 channels in a HEK293 cell expression system and in native mouse aorta endothelial cells. J
Biol Chem 277: 47044–47051. PMID: 12354759
44.
Xu H, Ramsey IS, Kotecha SA, Moran MM, Chong JA, Lawson D, et al. (2002) TRPV3 is a calciumpermeable temperature-sensitive cation channel. Nature 418: 181–186. PMID: 12077604
45.
Grimm C, Kraft R, Sauerbruch S, Schultz G, Harteneck C (2003) Molecular and functional characterization of the melastatin-related cation channel TRPM3. J Biol Chem 278: 21493–21501. PMID:
12672799
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
18 / 22
TRP Channels in Hair Cell Mechanotransduction
46.
Heiner I, Eisfeld J, Halaszovich CR, Wehage E, Jungling E, Zitt C, et al. (2003) Expression profile of
the transient receptor potential (TRP) family in neutrophil granulocytes: evidence for currents through
long TRP channel 2 induced by ADP-ribose and NAD. Biochem J 371: 1045–1053. PMID: 12564954
47.
Hofmann T, Chubanov V, Gudermann T, Montell C (2003) TRPM5 is a voltage-modulated and Ca(2
+)-activated monovalent selective cation channel. Curr Biol 13: 1153–1158. PMID: 12842017
48.
Luo Y, Vassilev PM, Li X, Kawanabe Y, Zhou J (2003) Native polycystin 2 functions as a plasma membrane Ca2+-permeable cation channel in renal epithelia. Mol Cell Biol 23: 2600–2607. PMID:
12640140
49.
Nilius B, Prenen J, Droogmans G, Voets T, Vennekens R, Freichel M, et al. (2003) Voltage dependence of the Ca2+-activated cation channel TRPM4. J Biol Chem 278: 30813–30820. PMID:
12799367
50.
Prawitt D, Monteilh-Zoller MK, Brixel L, Spangenberg C, Zabel B, Fleig A, et al. (2003) TRPM5 is a
transient Ca2+-activated cation channel responding to rapid changes in [Ca2+]i. Proc Natl Acad Sci U
S A 100: 15166–15171. PMID: 14634208
51.
Chung MK, Lee H, Mizuno A, Suzuki M, Caterina MJ (2004) 2-aminoethoxydiphenyl borate activates
and sensitizes the heat-gated ion channel TRPV3. J Neurosci 24: 5177–5182. PMID: 15175387
52.
LaPlante JM, Ye CP, Quinn SJ, Goldin E, Brown EM, Slaugenhaupt SA, et al. (2004) Functional links
between mucolipin-1 and Ca2+-dependent membrane trafficking in mucolipidosis IV. Biochem Biophys Res Commun 322: 1384–1391. PMID: 15336987
53.
Launay P, Cheng H, Srivatsan S, Penner R, Fleig A, Kinet JP (2004) TRPM4 regulates calcium oscillations after T cell activation. Science 306: 1374–1377. PMID: 15550671
54.
Shi J, Mori E, Mori Y, Mori M, Li J, Ito Y, et al. (2004) Multiple regulation by calcium of murine homologues of transient receptor potential proteins TRPC6 and TRPC7 expressed in HEK293 cells. J Physiol 561: 415–432. PMID: 15579537
55.
Grimm C, Kraft R, Schultz G, Harteneck C (2005) Activation of the melastatin-related cation channel
TRPM3 by D-erythro-sphingosine [corrected]. Mol Pharmacol 67: 798–805. PMID: 15550678
56.
Nagata K, Duggan A, Kumar G, Garcia-Anoveros J (2005) Nociceptor and hair cell transducer properties of TRPA1, a channel for pain and hearing. J Neurosci 25: 4052–4061. PMID: 15843607
57.
Nilius B, Prenen J, Janssens A, Owsianik G, Wang C, Zhu MX, et al. (2005) The selectivity filter of the
cation channel TRPM4. J Biol Chem 280: 22899–22906. PMID: 15845551
58.
Oberwinkler J, Lis A, Giehl KM, Flockerzi V, Philipp SE (2005) Alternative splicing switches the divalent cation selectivity of TRPM3 channels. J Biol Chem 280: 22540–22548. PMID: 15824111
59.
Li M, Jiang J, Yue L (2006) Functional characterization of homo- and heteromeric channel kinases
TRPM6 and TRPM7. J Gen Physiol 127: 525–537. PMID: 16636202
60.
Togashi K, Hara Y, Tominaga T, Higashi T, Konishi Y, Mori Y, et al. (2006) TRPM2 activation by cyclic
ADP-ribose at body temperature is involved in insulin secretion. Embo J 25: 1804–1815. PMID:
16601673
61.
Grimm C, Cuajungco MP, van Aken AF, Schnee M, Jors S, Kros CJ, et al. (2007) A helix-breaking
mutation in TRPML3 leads to constitutive activity underlying deafness in the varitint-waddler mouse.
Proc Natl Acad Sci U S A 104: 19583–19588. PMID: 18048323
62.
Xu H, Delling M, Li L, Dong X, Clapham DE (2007) Activating mutation in a mucolipin transient receptor potential channel leads to melanocyte loss in varitint-waddler mice. Proc Natl Acad Sci U S A 104:
18321–18326. PMID: 17989217
63.
Kim HJ, Li Q, Tjon-Kon-Sang S, So I, Kiselyov K, Soyombo AA, et al. (2008) A novel mode of
TRPML3 regulation by extracytosolic pH absent in the varitint-waddler phenotype. Embo J 27: 1197–
1205. doi: 10.1038/emboj.2008.56 PMID: 18369318
64.
Nagata K, Zheng L, Madathany T, Castiglioni AJ, Bartles JR, Garcia-Anoveros J (2008) The varitintwaddler (Va) deafness mutation in TRPML3 generates constitutive, inward rectifying currents and
causes cell degeneration. Proc Natl Acad Sci U S A 105: 353–358. PMID: 18162548
65.
Bobkov YV, Corey EA, Ache BW (2011) The pore properties of human nociceptor channel TRPA1
evaluated in single channel recordings. Biochim Biophys Acta 1808: 1120–1128. doi: 10.1016/j.
bbamem.2010.12.024 PMID: 21195050
66.
Zhang X, Li X, Xu H (2012) Phosphoinositide isoforms determine compartment-specific ion channel
activity. Proc Natl Acad Sci U S A 109: 11384–11389. doi: 10.1073/pnas.1202194109 PMID:
22733759
67.
DeCaen PG, Delling M, Vien TN, Clapham DE (2013) Direct recording and molecular identification of
the calcium channel of primary cilia. Nature 504: 315–318. doi: 10.1038/nature12832 PMID:
24336289
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
19 / 22
TRP Channels in Hair Cell Mechanotransduction
68.
Clapham DE, DeCaen P, Carvacho I, Chaudhuri D, Doerner JF, Julius D, et al. (2016) IUPHAR/BPS
Guide to PHARMACOLOGY. http://wwwguidetopharmacologyorg/GRAC/FamilyDisplayForward?
familyId=78.
69.
Liu H, Pecka JL, Zhang Q, Soukup GA, Beisel KW, He DZ (2014) Characterization of transcriptomes
of cochlear inner and outer hair cells. J Neurosci 34: 11085–11095. doi: 10.1523/JNEUROSCI.169014.2014 PMID: 25122905
70.
Kwan KY, Allchorne AJ, Vollrath MA, Christensen AP, Zhang DS, Woolf CJ, et al. (2006) TRPA1 contributes to cold, mechanical, and chemical nociception but is not essential for hair-cell transduction.
Neuron 50: 277–289. PMID: 16630838
71.
Sexton JE, Desmonds T, Quick K, Taylor R, Abramowitz J, Forge A, et al. (2016) The contribution of
TRPC1, TRPC3, TRPC5 and TRPC6 to touch and hearing. Neurosci Lett 610: 36–42. doi: 10.1016/j.
neulet.2015.10.052 PMID: 26520460
72.
Tabuchi K, Suzuki M, Mizuno A, Hara A (2005) Hearing impairment in TRPV4 knockout mice. Neurosci Lett 382: 304–308. PMID: 15925108
73.
Gerka-Stuyt J, Au A, Peachey NS, Alagramam KN (2013) Transient receptor potential melastatin 1: a
hair cell transduction channel candidate. PLoS One 8: e77213. doi: 10.1371/journal.pone.0077213
PMID: 24146970
74.
Alagramam KN, Goodyear RJ, Geng R, Furness DN, van Aken AF, Marcotti W, et al. (2011) Mutations
in protocadherin 15 and cadherin 23 affect tip links and mechanotransduction in mammalian sensory
hair cells. PLoS One 6: e19183. doi: 10.1371/journal.pone.0019183 PMID: 21532990
75.
Yang H, Gan J, Xie X, Deng M, Feng L, Chen X, et al. (2010) Gfi1-Cre knock-in mouse line: A tool for
inner ear hair cell-specific gene deletion. Genesis 48: 400–406. doi: 10.1002/dvg.20632 PMID:
20533399
76.
Meyers JR, MacDonald RB, Duggan A, Lenzi D, Standaert DG, Corwin JT, et al. (2003) Lighting up
the senses: FM1-43 loading of sensory cells through nonselective ion channels. J Neurosci 23:
4054–4065. PMID: 12764092
77.
Yao G, Luo C, Harvey M, Wu M, Schreiber TH, Du Y, et al. (2015) Disruption of Polycystin-L causes
Hippocampal Hyperexcitability. Hum Mol Genet.
78.
Tsiokas L, Kim E, Arnould T, Sukhatme VP, Walz G (1997) Homo- and heterodimeric interactions
between the gene products of PKD1 and PKD2. Proc Natl Acad Sci U S A 94: 6965–6970. PMID:
9192675
79.
Newby LJ, Streets AJ, Zhao Y, Harris PC, Ward CJ, Ong AC (2002) Identification, characterization,
and localization of a novel kidney polycystin-1-polycystin-2 complex. J Biol Chem 277: 20763–
20773. PMID: 11901144
80.
Murakami M, Ohba T, Xu F, Shida S, Satoh E, Ono K, et al. (2005) Genomic organization and functional analysis of murine PKD2L1. J Biol Chem 280: 5626–5635. PMID: 15548533
81.
Ishimaru Y, Katano Y, Yamamoto K, Akiba M, Misaka T, Roberts RW, et al. (2010) Interaction
between PKD1L3 and PKD2L1 through their transmembrane domains is required for localization of
PKD2L1 at taste pores in taste cells of circumvallate and foliate papillae. Faseb J 24: 4058–4067. doi:
10.1096/fj.10-162925 PMID: 20538909
82.
Matei V, Pauley S, Kaing S, Rowitch D, Beisel KW, Morris K, et al. (2005) Smaller inner ear sensory
epithelia in Neurog 1 null mice are related to earlier hair cell cycle exit. Dev Dyn 234: 633–650. PMID:
16145671
83.
Garcia-Gonzalez MA, Outeda P, Zhou Q, Zhou F, Menezes LF, Qian F, et al. (2010) Pkd1 and Pkd2
are required for normal placental development. PLoS One 5.
84.
Huang AL, Chen X, Hoon MA, Chandrashekar J, Guo W, Trankner D, et al. (2006) The cells and logic
for mammalian sour taste detection. Nature 442: 934–938. PMID: 16929298
85.
Ishimaru Y, Inada H, Kubota M, Zhuang H, Tominaga M, Matsunami H (2006) Transient receptor
potential family members PKD1L3 and PKD2L1 form a candidate sour taste receptor. Proc Natl Acad
Sci U S A 103: 12569–12574. PMID: 16891422
86.
Ye W, Chang RB, Bushman JD, Tu YH, Mulhall EM, Wilson CE, et al. (2016) The K+ channel KIR2.1
functions in tandem with proton influx to mediate sour taste transduction. Proc Natl Acad Sci U S A
113: E229–238. doi: 10.1073/pnas.1514282112 PMID: 26627720
87.
Nelson TM, Lopezjimenez ND, Tessarollo L, Inoue M, Bachmanov AA, Sullivan SL (2010) Taste function in mice with a targeted mutation of the pkd1l3 gene. Chem Senses 35: 565–577. doi: 10.1093/
chemse/bjq070 PMID: 20605874
88.
Eijkelkamp N, Quick K, Wood JN (2013) Transient receptor potential channels and mechanosensation. Annu Rev Neurosci 36: 519–546. doi: 10.1146/annurev-neuro-062012-170412 PMID:
23750514
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
20 / 22
TRP Channels in Hair Cell Mechanotransduction
89.
Lin SY, Corey DP (2005) TRP channels in mechanosensation. Curr Opin Neurobiol 15: 350–357.
PMID: 15922584
90.
Christensen AP, Corey DP (2007) TRP channels in mechanosensation: direct or indirect activation?
Nat Rev Neurosci 8: 510–521. PMID: 17585304
91.
Di Palma F, Belyantseva IA, Kim HJ, Vogt TF, Kachar B, Noben-Trauth K (2002) Mutations in Mcoln3
associated with deafness and pigmentation defects in varitint-waddler (Va) mice. Proc Natl Acad Sci
U S A 99: 14994–14999. PMID: 12403827
92.
van Aken AF, Atiba-Davies M, Marcotti W, Goodyear RJ, Bryant JE, Richardson GP, et al. (2008)
TRPML3 mutations cause impaired mechano-electrical transduction and depolarization by an inwardrectifier cation current in auditory hair cells of varitint-waddler mice. J Physiol 586: 5403–5418. doi:
10.1113/jphysiol.2008.156992 PMID: 18801844
93.
Jors S, Grimm C, Becker L, Heller S (2010) Genetic inactivation of Trpml3 does not lead to hearing
and vestibular impairment in mice. PLoS One 5: e14317. doi: 10.1371/journal.pone.0014317 PMID:
21179200
94.
Wang W, Zhang X, Gao Q, Xu H (2014) TRPML1: an ion channel in the lysosome. Handb Exp Pharmacol 222: 631–645. doi: 10.1007/978-3-642-54215-2_24 PMID: 24756723
95.
Hoenderop JG, van Leeuwen JP, van der Eerden BC, Kersten FF, van der Kemp AW, Merillat AM,
et al. (2003) Renal Ca2+ wasting, hyperabsorption, and reduced bone thickness in mice lacking
TRPV5. J Clin Invest 112: 1906–1914. PMID: 14679186
96.
Delling M, DeCaen PG, Doerner JF, Febvay S, Clapham DE (2013) Primary cilia are specialized calcium signalling organelles. Nature 504: 311–314. doi: 10.1038/nature12833 PMID: 24336288
97.
Nauli SM, Alenghat FJ, Luo Y, Williams E, Vassilev P, Li X, et al. (2003) Polycystins 1 and 2 mediate
mechanosensation in the primary cilium of kidney cells. Nat Genet 33: 129–137. PMID: 12514735
98.
Kottgen M, Buchholz B, Garcia-Gonzalez MA, Kotsis F, Fu X, Doerken M, et al. (2008) TRPP2 and
TRPV4 form a polymodal sensory channel complex. J Cell Biol 182: 437–447. doi: 10.1083/jcb.
200805124 PMID: 18695040
99.
Du J, Ma X, Shen B, Huang Y, Birnbaumer L, Yao X (2014) TRPV4, TRPC1, and TRPP2 assemble to
form a flow-sensitive heteromeric channel. Faseb J 28: 4677–4685. doi: 10.1096/fj.14-251652 PMID:
25114176
100.
Delling M, Indzhykulian AA, Liu X, Li Y, Xie T, Corey DP, et al. (2016) Primary cilia are not calciumresponsive mechanosensors. Nature 531: 656–660. doi: 10.1038/nature17426 PMID: 27007841
101.
Shin JB, Krey JF, Hassan A, Metlagel Z, Tauscher AN, Pagana JM, et al. (2013) Molecular architecture of the chick vestibular hair bundle. Nat Neurosci 16: 365–374. doi: 10.1038/nn.3312 PMID:
23334578
102.
LopezJimenez ND, Cavenagh MM, Sainz E, Cruz-Ithier MA, Battey JF, Sullivan SL (2006) Two members of the TRPP family of ion channels, Pkd1l3 and Pkd2l1, are co-expressed in a subset of taste
receptor cells. J Neurochem 98: 68–77. PMID: 16805797
103.
Ishii S, Misaka T, Kishi M, Kaga T, Ishimaru Y, Abe K (2009) Acetic acid activates PKD1L3-PKD2L1
channel—a candidate sour taste receptor. Biochem Biophys Res Commun 385: 346–350. doi: 10.
1016/j.bbrc.2009.05.069 PMID: 19464260
104.
Steigelman KA, Lelli A, Wu X, Gao J, Lin S, Piontek K, et al. (2011) Polycystin-1 is required for stereocilia structure but not for mechanotransduction in inner ear hair cells. J Neurosci 31: 12241–12250.
doi: 10.1523/JNEUROSCI.6531-10.2011 PMID: 21865467
105.
Yu Y, Ulbrich MH, Li MH, Buraei Z, Chen XZ, Ong AC, et al. (2009) Structural and molecular basis of
the assembly of the TRPP2/PKD1 complex. Proc Natl Acad Sci U S A 106: 11558–11563. doi: 10.
1073/pnas.0903684106 PMID: 19556541
106.
Corey DP, Garcia-Anoveros J, Holt JR, Kwan KY, Lin SY, Vollrath MA, et al. (2004) TRPA1 is a candidate for the mechanosensitive transduction channel of vertebrate hair cells. Nature 432: 723–730.
PMID: 15483558
107.
Liman ER, Corey DP, Dulac C (1999) TRP2: a candidate transduction channel for mammalian pheromone sensory signaling. Proc Natl Acad Sci U S A 96: 5791–5796. PMID: 10318963
108.
Stowers L, Holy TE, Meister M, Dulac C, Koentges G (2002) Loss of sex discrimination and malemale aggression in mice deficient for TRP2. Science 295: 1493–1500. PMID: 11823606
109.
Liman ER, Innan H (2003) Relaxed selective pressure on an essential component of pheromone
transduction in primate evolution. Proc Natl Acad Sci U S A 100: 3328–3332. PMID: 12631698
110.
Low SE, Amburgey K, Horstick E, Linsley J, Sprague SM, Cui WW, et al. (2011) TRPM7 is required
within zebrafish sensory neurons for the activation of touch-evoked escape behaviors. J Neurosci 31:
11633–11644. doi: 10.1523/JNEUROSCI.4950-10.2011 PMID: 21832193
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
21 / 22
TRP Channels in Hair Cell Mechanotransduction
111.
Liedtke W, Tobin DM, Bargmann CI, Friedman JM (2003) Mammalian TRPV4 (VR-OAC) directs
behavioral responses to osmotic and mechanical stimuli in Caenorhabditis elegans. Proc Natl Acad
Sci U S A 100 Suppl 2: 14531–14536. PMID: 14581619
112.
Forge A, Nevill G, Zajic G, Wright A (1992) Scanning electron microscopy of the mammalian organ of
Corti: assessment of preparative procedures. Scanning Microsc 6: 521–534; discussion 534–525.
PMID: 1462137
113.
Indzhykulian AA, Stepanyan R, Nelina A, Spinelli KJ, Ahmed ZM, Belyantseva IA, et al. (2013) Molecular remodeling of tip links underlies mechanosensory regeneration in auditory hair cells. PLoS Biol
11: e1001583. doi: 10.1371/journal.pbio.1001583 PMID: 23776407
PLOS ONE | DOI:10.1371/journal.pone.0155577 May 19, 2016
22 / 22
Download